Summary
Major progress in the analysis of human immune responses to cancer has been made through the molecular characterization of human tumour antigens. The development of therapeutic strategies for eliciting immune‐mediated rejection of tumours has accelerated due to the elucidation of the molecular basis for tumour cell recognition and destruction by immune cells. Of the various human tumour antigens defined to date in ovarian cancer, the cancer‐testis (CT) family of antigens have been studied extensively preclinically and clinically because of their testis‐restricted expression in normal tissues and ability to elicit robust immune responses. Recent developments in cancer sequencing technologies offer a unique opportunity to identify tumour mutations with the highest likelihood of being expressed and recognized by the immune system. Such mutations, or neoantigens, could potentially serve as specific immune targets for T‐cell‐mediated destruction of cancer cells. This review will highlight current work in selecting tumour rejection antigens in ovarian cancer for improving the efficacy of immunotherapy.
Keywords: antigen, immunotherapy, next‐generation sequencing, ovarian cancer
Introduction
The concept of harnessing the immune system to fight neoplasms was first pioneered by William Coley in 1890. Upon injecting streptococcal organisms into patients with bone sarcomas,1 Coley observed a significant reduction in tumour size. He attributed this response to the activation of the immune system to bacterial infection2 and so provided the earliest evidence for immune‐mediated regression of established tumours. Tremendous progress in developing immunotherapy for antitumour therapy has been made since Coley's observation, such that immunotherapy has emerged as the most promising cancer treatment since the development of chemotherapy in the 1940s. Part of this success can be attributed to the potential of the immune system to exhibit exquisite specificity for cancer cells while simultaneously minimizing adverse effects.3 The role of immune cells, especially tumour‐reactive T cells, in eradicating established tumour has been reported in murine models4, 5, 6 and in numerous clinical trials.7, 8, 9 The dramatic clinical responses in certain types of solid cancers treated with monoclonal antibodies targeting immune checkpoint pathways has spurred the recent popularity of using the immune system to control unchecked tumour growth. US Food and Drug Administration (FDA) approval of checkpoint inhibitors, anti‐cytotoxic T lymphocyte antigen 4, anti‐programmed death 1 and sipuleucel‐T for metastatic prostate cancer10, 11 suggest that these promising results may expand the breadth of diseases, such as ovary, liver, colon and lung cancer, that will similarly benefit from immune‐based therapies.12 As a result of this boom in immunotherapy research, both the FDA and the European Medicines Agency (EMA) approved a number of immunotherapeutic agents for the treatment of bladder, prostate, lung, head and neck cancers, melanoma, lymphoma and synovial sarcoma.12
Immunotherapy in ovarian cancer
Ovarian cancer (OC) is the seventh most common cancer in the USA and the principal cause of death from gynaecological malignancies in the world.13 Early‐stage OC is mainly asymptomatic so OC is often diagnosed at a late stage when the disease has already spread throughout the peritoneal cavity, often accompanied by malignant ascites. Approximately 75% of women diagnosed with OC are already in stage II–IV and current frontline management of the disease includes debulking surgery and chemotherapy.14 Despite the short‐term benefit of cytotoxic agents as first‐line chemotherapy, only 30% of women with late‐stage disease survive more than 5 years post‐diagnosis.
Initial studies in women with ovarian cancer observed immune cell infiltrates within the tumour, but these studies did not establish a definite link to clinical outcomes.15 In order to evaluate the role of the immune system in ovarian malignancies, a 2003 study quantified the immune infiltrates in 186 advanced‐stage ovarian carcinomas and detected tumour‐infiltrating lymphocytes (TILs) in 54·8% of samples, with TIL infiltration correlating with longer disease progression and survival.16 Later work on 117 OC samples by Sato et al. analysed using immunohistochemistry described improved clinical outcome for patients with high versus low ratio of intratumoral CD8+ and regulatory T (Treg) cells versus patients with a subpopulation of TILs, CD8+ T cells strongly correlating with prolonged survival.17 These results were later confirmed by Hwang et al., who performed a meta‐analysis of 1815 women with OC and corroborated the correlation between the presence of TILs and increased overall survival.18 In addition to T cells, the tumour microenvironment contains additional subsets of immune cells exhibiting a capacity to suppress antitumour immunity and rather promote tumour growth. High infiltration of CD4+ CD25+ FOXP3+ Treg cells in OC tumours was reported to promote tumour progression by suppressing production of interferon‐γ and interleukin‐2 in antitumour effector T cells. Homing of Treg cells within the tumour is elicited by the production of the C‐C motif chemokine 22 (CCL22) by tumour cells and tumour‐associated macrophages (TAMs) type 2 (M2).19 In addition to CCL22, M2 macrophages also express B7‐H4, which decreases the proliferation and activation of tumour specific T cells.20, 21, 22 Blocking CCL2 and B7‐H4 with monoclonal antibodies depletes M2 macrophages at the tumour site through inhibition of the macrophage colony‐stimulating factor. Anti‐macrophage colony‐stimulating factor monoclonal antibodies have proven effective in preclinical tumour models by increasing the CD8+ T cell to CD4+ T cell ratio at the tumour site and has entered clinical trials for advanced solid tumours including OC in 2011 (NCT01494688). The study attributed tumour shrinkage in patients with tenosynovial giant cell tumours to a reduction in TAMs.23 These studies indicate that either depletion of immunosuppressive cellular elements (i.e. Treg cells or TAMs) in the tumour can enhance survival in advanced solid tumours by promoting tumour homing of antitumour CD8+ T cells. These data suggest that modulation of suppressive elements in the tumour microenvironment holds a great promise for improving clinical outcome of OC patients.
Endogenous tumour antigens
The aberrant behaviour of tumour cells can be attributed to a multitudinous array of molecular alterations including altered gene expression and genetic mutations. In early efforts aimed to profile the immunopeptidome, defined as the collection of all immunogenic peptides present in a sample, cDNA libraries were transfected into COS or 293 cells expressing appropriate major histocompatibility complex (MHC) molecules and reactive cytotoxic T lymphocytes clones were identified after co‐culturing experiments.24, 25, 26 Although this approach identified numerous potential antigens, cross‐reactivity with inert peptides was also reported. With the goal of obtaining antigen specificity for tumour cells, peptides bound to the MHC or on the tumour cell surface are eluted and separated by chromatography. Eluted peptides are then tested for T‐cell recognition after being pulsed on human leucocyte antigen (HLA) matched antigen‐presenting cells and co‐cultured with T cells. Reactive peptides can also be sequenced by Edman degradation, in which amino acids are sequentially labelled and cleaved before identification,27 and ultimately mapped onto the encoding genes. This step is critical as identification of the parent gene allows epitopes to be stratified into whether they are derived from wild‐type genes or tumour mutation‐specific genes. Epitopes derived from mutations may therefore elicit responses from T cells that have escaped central tolerance mechanisms commonly associated with tolerance to ubiquitous normal expressed epitopes. Further characterization of the immunopeptidome and tumour‐associated neoantigens has become an active area of analysis to generate tumour‐specific T‐cell immunity.28, 29, 30
Antigens are generally classified into self and non‐self, with tumour antigens divided into self and mutated‐self. Mutated‐self antigens include neoantigens (described below) while self‐antigens include the cancer testis (CT) antigens, which are expressed during fetal development, have a limited expression on normal cells and are expressed at higher levels in cancerous cells. Analysis of the pattern of expression of 162 CT antigens across 53 normal samples from GTeX and 31 tumour samples from The Cancer Genome Atlas (TCGA) revealed a strikingly contrasting profile. TCGA tumour samples had a heterogeneous expression of all CT antigens, all while normal tissues beside testis had minimal or absent CT expression (Fig. 1). These results strongly suggest that CT antigens can be exquisite candidates for clinical applications as immune targets in OC and other cancers. Furthermore, as transcription of CT is epigenetically regulated,31, 32, 33 there are opportunities to reinstate CT expression by treating patients with DNA methyltransferase inhibitors, such as decitabine‐derived drugs.
Figure 1.

Expression pattern of 162 cancer‐testis (CT) genes across normal tissues from GTeX and patient tumour samples from TCGA. RNASeq data were obtained from GTeX (normal tissues) or TCGA PANCancer study (tumours). The median expression per each CT gene was calculated across all patients in a specific tissue. Each cell in the heatmap indicates the median expression of a CT gene in the tissue indicated at the bottom of the figure. Red cells indicate high expression and black cells low expression levels.
The CT family of tumour antigens encompasses > 250 members and the most commonly studied include: New York oesophageal‐1 (NY‐ESO‐1), which is expressed in ovarian carcinoma, melanoma, sarcoma, breast carcinoma, bladder carcinoma, prostate cancer and hepatocellular carcinoma;34, 35 B melanoma antigen (BAGE), which is expressed in melanoma, bladder carcinoma, mammary carcinoma, head and neck squamous cell carcinoma, non‐small cell lung carcinoma36; melanoma‐associated antigen‐A (MAGE‐A), which is expressed in epithelial carcinomas and germ cells including cutaneous squamous cell carcinomas, oesophageal, head and neck, bladder urothelial, cervical/anal, lung, lung adenocarcinomas, ovarian, endometrial, lung small cell, gastric adenocarcinomas, breast mucinous, hepatocellular, breast infiltrating ductal, colorectal adenocarcinomas, cholangiocarcinomas, thymic and mesotheliomas.37 MAGE‐1 or melanoma antigen‐1 was the first tumour antigen discovered more than two decades ago from human melanoma cell lines.38, 39, 40, 41 Targeting CT antigens in situations where CT genes are expressed in non‐carcinogenic tissues, led to severe adverse effects, including acute inflammation in the brain that caused neuronal cell degradation and myocardial damage.42, 43 Odunsi et al. reported higher expression of the CT antigens, NY‐ESO‐1 and L antigen family member 1 (LAGE‐1) in women with epithelial ovarian cancer and successfully targeted them in these patients.44, 45 NY‐ESO‐1 antigens are recognized by both helper CD4+ T cells as well as CD8+ T cells and these cells were found to be elevated in patients after a course of at least five vaccinations of NY‐ESO‐1 peptide at 3‐week intervals. Vaccine‐elicited CD4+ and CD8+ T cells were shown to recognize NY‐ESO‐1‐expressing tumour targets and were detectable in some patients up to 12 months after immunization.44
Cancer‐testis antigens have also been exploited as targets for adoptive T‐cell therapy with various degrees of clinical response. Adoptive T‐cell therapy in 36 melanoma patients using autologous peripheral blood lymphocytes transduced with a Melan A1 specific T‐cell receptor (TCR) mediated significant tumour regression while being associated with adverse toxicity due to recognition of Melan A1 in melanocytes in the skin, eyes and ears.46 Carcinoembryonic antigen is expressed in gastrointestinal tissue of fetus before birth and later appears at low levels in the blood of healthy individuals. Carcinoembryonic antigen was tested as potential tumour‐associated antigen in colorectal adenocarcinoma patients. Autologous T lymphocytes of three patients were genetically engineered to express murine TCR directed against human carcinoembryonic antigen. All patients experienced a profound decrease in serum carcinoembryonic antigen levels with one patient showing a drastic reduction in cancer metastasis to the liver and lung. However, severe colitis was observed in all patients representing dose‐related toxicity of engineered T cells, having TCRs raised in HLA transgenic mice.47 These antigens including cancer testis antigens, differentiated self‐antigens and over‐expressed antigens have shown limited success in the clinical setting.48 These studies strongly indicate the need for tumour‐specific antigens, which are not expressed in normal cells and could therefore provide optimal specificity for malignant cells.
Clinical trials of T‐cell therapy targeting antigens in ovarian cancer
Engineering autologous T cells with TCRs that recognize tumour‐associated epitope is a promising strategy for treating cancer patients through generating a large number of therapeutic T cells with the potential to expand in vivo and exert potent antitumour activity. T‐cell therapies have been implemented with various degrees of success in OC and are currently used in Phase 1 and Phase 2 clinical trials (Table 1). Here we offer a brief description of OC trials that implement immune techniques to bolster antigen‐specific tumour rejection.
Table 1.
Clinical trials of T‐cell therapy targeting different antigens in ovarian cancer; neoantigens elicit antitumour immunity
| Antigen type | Target | Clinical trials ID (Phase) |
|---|---|---|
| CT antigen | NY‐ESO1 | NCT03159585 (Phase 1) |
| NCT01567891 (Phase 1/2) | ||
| NCT03017131 (Phase 1) | ||
| NCT00101257 (Phase 1) | ||
| NCT02457650 (Phase 1) | ||
| NCT02166905 (Phase 1/2) | ||
| CT antigen | MAGE‐A4 | NCT03132922 (Phase 1) |
| CT antigen | CEA | NCT01212887 (Phase 1) |
| Oncogenic antigen | MUC16 | NCT02498912 (Phase1) |
| Oncogenic antigen | HER2 | NCT00194714 (Phase 1/2) |
| NCT00228358 (Phase 1) | ||
| Oncogenic antigen | WT1 | NCT00562640 (Phase 1) |
| Oncogenic antigen | Mesothelin | NCT02580747 (Phase 1) |
| NCT03054298 (Phase 1) | ||
| NCT01583686 (Phase 1/2) | ||
| NCT02159716 (Phase 1) | ||
| Oncogenic antigen | Prominin‐1 | NCT02541370 (Phase 1) |
| Neoantigens | Mutated genes | NCT03412877 (Phase 2) |
| NCT02876510 (Phase 1) |
The human epidermal growth factor 2 (HER2) is a proto‐oncogene encoding for the tyrosine kinase receptor that modulates cell growth and survival. Altered HER2 levels can be found in gynaecological malignancies49, 50, 51 with HER2 over‐expression correlating with increased risk of progression and shorter survival in OC patients with FIGO stage I and II.52 Moreover, HER2 proteins expressed in OC were shown to bind to and activate chimeric antigen receptor (CAR) T cells with a potent antitumour activity against tumour cells from ascites, solid tumours and established HER2‐expressing OC cell line.49 Engineered cytotoxic T cells expressing HER2‐specific TCR have been used to recognize HER2 epitopes on OC cells and led to delayed progression of human tumour in xenograft models.53
Another target antigen with promising results as clinical target for OC patients is NY‐ESO‐1. NY‐ESO‐1 is a CT antigen that, as with other CT antigens, is expressed mainly in the testis with little or no expression in normal tissues (Fig. 1). However, its expression is reinstated in cancer cells so that serum antibodies against NY‐ESO‐1 have been detected in multiple malignancies including colorectal,54 gastric,55 oesophageal56 and ovarian cancers.57, 58 Although detected, NY‐ESO‐1 expression in the tumour can be heterogeneous due to the epigenetic mechanisms of transcriptional silencing, such as DNA methylation or histone modifications, therefore, alternative approaches that involve the use of DNA demethylating or histone deacetylase inhibitor agents have been used to restore NY‐ESO‐1 levels.59, 60, 61, 62 NY‐ESO‐1‐specific T cells were readily detected in the TILs and tumour‐associated lymphocytes from 71% of NY‐ESO‐1‐seropositive OC patients, suggesting that patients can mount an autologous adaptive immune response against NY‐ESO‐1‐expressing tumour cells.63 Therapeutically, TCR engineered T cells specific against NY‐ESO‐1 have successfully generated an antitumour response in 11 out of 18 patients with synovial cell carcinoma and 11 out of 20 patients with metastatic melanoma in a Phase 1 clinical trial with no signs of toxicity.25, 64 Based on these promising results, several studies targeting NY‐ESO‐1 alone or in combination in seropositive OC patients currently in Phase 1 and Phase 2 clinical trials are listed in Table 1.
The α folate receptor 1 (FR1) is a 38 000 mol wt glycosylinositol anchored protein encoded by the gene FOLR1, responsible for the uptake of folate required for cell survival, proliferation and gene expression. Tumour‐restricted expression of FOLR1 is reported in 90% of OC patients and its expression correlates with poor survival.65, 66 The effect of targeting FR1 using Bi‐specific T‐cell engaging (BiTE) antibodies composed of anti‐CD3 and anti‐FR1 antibodies was evaluated for a phase II study in 26 OC patients with limited intraperitoneal disease, seven of which showed complete or partial intraperitoneal response with strict surgicopathological evaluation.67 CAR T cells engineered with anti‐FOLR1‐scFv and IgE Fc receptor signalling domain failed to establish tumour control in a Phase 1 clinical trial after encouraging in vitro results.68 The authors hypothesized that low expression of transgenic CAR, limited T‐cell persistence and inadequate tumour homing could account for the negative results.69 An enhancement over the previous FOLR1‐targeting CAR T cells came with the addition of CD137 and CD27 genes into the CAR construct, with consequent enhanced persistence of T cells at the tumour site and tumour regression in preclinical settings.70
Mesothelin (MSLN) is a GPI‐anchored protein expressed in 60–70% of epithelial OC patients.71, 72 In OC, spontaneous immune responses against MSLN have been detected and high levels of MSLN correlates with poor prognosis.73 CAR T cells consisting of CD28 domain, 4‐1BB and MSLN‐specific TCR have been tested preclinically in mouse OC models expressing mesothelin, resulting in a marked reduction of tumour or complete regression.74 Safety and feasibility studies of MSLN‐specific CAR T cells (CART‐meso) containing the intracellular signalling domain of 4‐1BB and CD3ζ in six OC patients have demonstrated T cells homing to the tumour site and clearing of tumour cells in the pleural effusion of one patient.75 Although preliminary results of clinical trials with MSLN CAR T cells in OC patients were shown to be well tolerated with any off‐target toxicities, T cells engineered to express an MSLN‐specific TCR are still under investigation in OC patients despite showing promising antitumour response in preclinical models.76
MAGE‐A is a CT antigen encoded by the MAGE‐A gene located on chromosome Xq28.77 Although MAGE‐A transcription is silenced in normal tissues but expressed in male germ cells and placenta, it is expressed at detectable levels in a variety of cancers of diverse histological origins.78, 79 Expression of the MAGE‐A4 antigen was observed in 186 out of 399 OC patients and has been studied as an attractive immunotherapy target.80, 81 A recent Phase 1 clinical trial started recruiting HLA‐A*02‐positive patients to be treated with genetically engineered T cells targeting MAGE‐A4 in multiple solid tumours, including OC (Table 1).
The cancer antigen 125 (CA125) was identified in 1981 as a reference standard biomarker, whose expression is elevated in 70–80% of OC patients.82 CA125 is derived from the cleavage of a large transmembrane glycoprotein, MUC16.82, 83 As CA125 expression is limited to carcinogenic ovarian tissues, CAR T cells specific for MUC16 were developed and tested in preclinical models of OC bearing human MUC16+ tumours. Adoptive T‐cell therapy with these CAR T cells resulted in either complete eradication or delayed tumour progression.84 Further, CAR T cells targeting MUC16 were modified to secrete interleukin‐12 for enhancing in vivo persistence of these cells, they and are currently in Phase 1 clinical trials in women with recurrent OC with elevated MUC16 expression85 (Table 1).
Other endogenous antigens currently being tested in Phase 1 clinical trials as targets for T‐cell therapy in OC include prominin 1, a glycoprotein encoded by the PROM1 gene, and Wilms tumour protein 1 (WT1), an oncogenic protein that contributes to the malignant and invasive phenotypes of tumour (Table 1).
Growing evidence suggests that targeting tumour antigens with engineered T cells is a promising treatment opportunity for OC patients. However, engineered T cells redirected against a tumour‐associated antigen have a modest antitumour response in clinical trials, probably because of several limitations such as limitation of TCRs in recognizing only MHC‐bound antigens, intracellular loss or mutation of MHC expression, alterations in the antigen‐processing machinery and the presence of a suppressive tumour microenvironment.48, 86, 87 Furthermore, as antigen loss is a common mechanism of immune escape, redirecting T cells towards multiple antigens could overcome the tumour defence mechanisms and, if safe, offer unique therapeutic opportunities for OC patients.
Neoantigens elicit antitumour immunity
Advancement in algorithms to identify somatic mutations, coupled with computational modelling of the antigen processing and presentation machinery led to the identification of tumour‐specific mutated self‐antigens predicted to be immunogenic, namely neoantigens or neoepitopes. Neoantigens are derived from somatic missense mutations that alter the translated amino acid sequence within the expressed protein product. As normal cells do not harbour somatic mutations, they do not express the mutated final peptide product (or neoantigen), and the tumour‐restricted expression of the neoantigens makes them attractive targets for immunotherapy. The first approach exploiting mutated antigens to prime tumour regression was initiated by Mandelboim et al. in 1995, who utilized peptides derived from a 37 gap‐junction protein to vaccinate mice against 3LL‐D122 lung carcinoma and noticed a CD8+ T‐cell‐driven antitumour response.88
Currently a number of in silico prediction tools allow users to infer peptides immunogenicity modelled as affinity to the HLA/MHC complex and probability of being presented on the cell's surface. These tools are developed around flavours of machine learning algorithms that, starting on previous binding data for known peptide–MHC binding profiles, can predict the likelihood of a chosen peptide to bind to a specific MHC. The most comprehensive tools to predict immunogenicity from peptide sequences are listed in Table 2.
Table 2.
List of the available algorithms for inferring peptide binding to the MHC and immunogenicity
| Name | Website | MHC class | Algorithm | Reference |
|---|---|---|---|---|
| NetMHCpan | http://www.cbs.dtu.dk/services/NetMHCpan/ | I | ANN | 89, 90, 91 |
| NetMHC | http://www.cbs.dtu.dk/services/NetMHC/ | I | ANN | 92, 93 |
| NetMHCII | http://www.cbs.dtu.dk/services/NetMHCII/ | II | ANN | 94, 95 |
| PickPocket | http://www.cbs.dtu.dk/services/PickPocket/ | I | PSSM | 96 |
| PSSMHCPan | https://github.com/BGI2016/PSSMHCpan | I | PSSM | 97 |
| IEDB | http://tools.iedb.org/main/ | I, II | ANN, PSSM | 98 |
| mhcflurry | https://github.com/openvax/mhcflurry | I | KNN | 99 |
| SYFPEITHI | http://www.syfpeithi.de/ | I | ANN | 100 |
ANN, artificial neural network; KNN, keras neural network; PSSM, position‐specific scoring matrix.
Peptide products containing somatic mutation can be exquisitely discriminated against wild‐type peptides by T cells bearing neoantigen‐specific TCRs. Tumour‐specific neoantigens have an advantage over tumour‐associated antigens in that T cells expressing TCRs specific for neoantigens are not subjected to central tolerance mechanisms,101 and are therefore likely to be present at a higher frequency in the overall TCR repertoire compared with TCRs with specificity for tumour‐associated antigens. Studies have demonstrated that neoantigens can be recognized by cytotoxic T lymphocytes and can be used in prophylactic or therapeutic settings102 and neoantigen‐reactive T‐cell clones from tumour tissues and peripheral blood lymphocytes have been identified in patients with different malignancies,103, 104, 105, 106, 107 indicating that autologous reactivity to mutated peptides is a shared mechanism of antitumour immunity. On the other hand, these studies suggest that only a handful of non‐synonymous mutations can generate neoantigens in cancer cells that will successfully lead to tumour rejection. In a recent study, Hartmaier et al. performed targeted sequencing across 63 220 tumours to identify shared neoantigens from patients bearing different malignancies and HLA types. The authors showed a remarkably minute overlap of predicted immunogenic neoantigens across patients,108 therefore strongly limiting the utilization of a single shared neoantigen ‘across the board’. The utilization of a selected panel of cancer‐related genes, might explain the limited number of shared antigens reported in the manuscript. A comprehensive evaluation of published WES/WGS data would offer a much broader panel of potential targets but still present the challenge of diluting neoantigen frequency throughout the whole genome; an alternative could be using the ratio of neoantigens per total number of coding base pairs analysed as proxy for the probability of identifying shared neoantigens across samples. However, although published pipelines suggest how to computationally predict neoantigens from sequencing data,30, 109, 110 only wet laboratory validation can confirm true HLA binding and T‐cell activation, hence, antitumour activity. An alternative approach to common next‐generation sequencing‐based studies exploits mass spectrometry techniques to identify mutated peptides while bound to MHC molecules on tumour cells.111, 112 Although this methodology captures a real‐life snapshot of the interface between immune system and tumour antigens, therefore bypassing the need to query immunogenicity prediction tools, it still requires validation experiments to confirm true positive calls. Validation in the forms of T‐cell activation measured as interferon‐γ production and CD8+ CD137+ T‐cell proliferation is a reliable in vitro readout of neoantigen recognition; however, it does not always translate into a strong in vivo activity. To understand the correlation between neoantigen frequency and T‐cell function, McGranahan et al. surveyed intratumour heterogeneity in parallel with neoantigen burden in melanoma and lung cancer patients undergoing checkpoint blockade therapy. Interestingly they demonstrated that patients with high clonal neoantigen burden and low intratumour heterogeneity present a highly inflamed microenvironment and positively respond to anti‐programmed death 1 and anti‐cytotoxic T lymphocyte antigen 4 therapy.113 These results indicate that understanding a patient's mutational landscape in the context of the tumour microenvironment can be greatly beneficial as a prognostic marker for the response to checkpoint blockade therapy. As this study focused on tumours with high mutational load (melanoma and lung cancer), it is still important to evaluate whether the same concept holds true for other cancer types that, like ovarian cancer, have an overall low mutational burden.
Future applications and directions
Advances in cancer immunogenomics technologies allow research and clinical investigators to profile with precision a patient's tumour, tumour microenvironment and circulating immunome. In parallel, as the complexity of immuno‐oncology multidimensional data sets increases, the interpretation of these results for clinical intervention remains limited, hence results become more descriptive than translational. The goal of developing truly personalized therapeutic approaches that leverage the patient's antigenome has been achieved in small clinical trials and, as described in this manuscript, numerous others have been approved. However, as precision immunotherapy fosters the skills to develop tailored therapies, the practical requirements of establishing a consistent, quick and reliable antigen identification and validation pipeline in every clinic is not yet doable. An accurate platform built on the interface between clinics, wet laboratories and bioinformaticians would be the perfect macroenvironment to foster proactive translational hubs that efficiently forge‐omics findings into clinical approaches. As few cancer centres offer this, the number of cancer patients that could benefit from tumour antigen‐based immunotherapies speaks volume about the need for efficient implementation of these therapeutic pipelines. Examples of ‘off‐the‐shelf’ coupled tumour antigens and clinically usable TCR sequences are limited to the KRAS G12D TCR developed by the Rosenberg's laboratory8, 114, 115 and identifying other pairs of antigen moieties and TCR sequences could offer great benefits for patients seeking passive immunotherapies as well as for developing vaccination strategies. Overall, our immune system has the potential to efficiently recognize a multitudinous array of antigens and efficiently target, and in some cases eradicate, tumour cells in patients. As opportunities to bolster the antitumour immune response grow, so does the potential to make a long lasting impact in patient care. The growing field of ‘integrative immunogenomics’ is therefore a fertile ground to study, understand and exploit the power of the immune system against cancer.
Disclosures
None to declare.
References
- 1. Nauts HC, Swift WE, Coley BL. The treatment of malignant tumors by bacterial toxins as developed by the late William B. Coley, M.D., reviewed in the light of modern research. Cancer Res 1946; 6:205–16. [PubMed] [Google Scholar]
- 2. McCarthy EF. The toxins of William B. Coley and the treatment of bone and soft‐tissue sarcomas. Iowa Orthop J 2006; 26:154–8. [PMC free article] [PubMed] [Google Scholar]
- 3. Alatrash G, Jakher H, Stafford PD, Mittendorf EA. Cancer immunotherapies, their safety and toxicity. Expert Opin Drug Saf 2013; 12:631–45. [DOI] [PubMed] [Google Scholar]
- 4. Melief CJ, Kast WM. T‐cell immunotherapy of cancer. Res Immunol 1991; 142:425–9. [DOI] [PubMed] [Google Scholar]
- 5. Greenberg PD. Adoptive T cell therapy of tumors: mechanisms operative in the recognition and elimination of tumor cells. Adv Immunol 1991; 49:281–355. [DOI] [PubMed] [Google Scholar]
- 6. Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME. Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Nat Rev Cancer 2008; 8:299–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Lu YC, Parker LL, Lu T, Zheng Z, Toomey MA, White DE et al Treatment of patients with metastatic cancer using a major histocompatibility complex class II‐restricted T‐cell receptor targeting the cancer germline antigen MAGE‐A3. J Clin Oncol 2017; 35:3322–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Tran E, Robbins PF, Lu YC, Prickett TD, Gartner JJ, Jia L et al T‐cell transfer therapy targeting mutant KRAS in cancer. N Engl J Med 2016; 375:2255–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Rapoport AP, Stadtmauer EA, Binder‐Scholl GK, Goloubeva O, Vogl DT, Lacey SF et al NY‐ESO‐1‐specific TCR‐engineered T cells mediate sustained antigen‐specific antitumor effects in myeloma. Nat Med 2015; 21:914–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Mellman I, Coukos G, Dranoff G. Cancer immunotherapy comes of age. Nature 2011; 480:480–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell 2015; 161:205–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Martin‐Liberal J, Ochoa de Olza M, Hierro C, Gros A, Rodon J, Tabernero J. The expanding role of immunotherapy. Cancer Treat Rev 2017; 54:74–86. [DOI] [PubMed] [Google Scholar]
- 13. Jayson GC, Kohn EC, Kitchener HC, Ledermann JA. Ovarian cancer. Lancet 2014; 384:1376–88. [DOI] [PubMed] [Google Scholar]
- 14. Herzog TJ. The current treatment of recurrent ovarian cancer. Curr Oncol Rep 2006; 8:448–54. [DOI] [PubMed] [Google Scholar]
- 15. Haskill S, Becker S, Fowler W, Walton L. Mononuclear‐cell infiltration in ovarian cancer. I. Inflammatory‐cell infiltrates from tumour and ascites material. Br J Cancer 1982; 45:728–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Zhang L, Conejo‐Garcia JR, Katsaros D, Gimotty PA, Massobrio M, Regnani G et al Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med 2003; 348:203–13. [DOI] [PubMed] [Google Scholar]
- 17. Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, Qian F et al Intraepithelial CD8+ tumor‐infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci USA 2005; 102:18538–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Hwang WT, Adams SF, Tahirovic E, Hagemann IS, Coukos G. Prognostic significance of tumor‐infiltrating T cells in ovarian cancer: a meta‐analysis. Gynecol Oncol 2012; 124:192–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P et al Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med 2004; 10:942–9. [DOI] [PubMed] [Google Scholar]
- 20. Sica GL, Choi IH, Zhu G, Tamada K, Wang SD, Tamura H et al B7‐H4, a molecule of the B7 family, negatively regulates T cell immunity. Immunity 2003; 18:849–61. [DOI] [PubMed] [Google Scholar]
- 21. Kryczek I, Zou L, Rodriguez P, Zhu G, Wei S, Mottram P et al B7‐H4 expression identifies a novel suppressive macrophage population in human ovarian carcinoma. J Exp Med 2006; 203:871–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Dangaj D, Lanitis E, Zhao A, Joshi S, Cheng Y, Sandaltzopoulos R et al Novel recombinant human b7‐h4 antibodies overcome tumoral immune escape to potentiate T‐cell antitumor responses. Cancer Res 2013; 73:4820–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Cassier PA, Gomez‐Roca CA, Italiano A, Cannarile M, Ries C, Brillouet A et al Phase 1 study of RG7155, a novel anti‐CSF1R antibody, in patients with locally advanced pigmented villonodular synovitis (PVNS). J Clin Oncol 2014; 32:10504. [Google Scholar]
- 24. Brichard V, Van Pel A, Wolfel T, Wolfel C, De Plaen E, Lethe B et al The tyrosinase gene codes for an antigen recognized by autologous cytolytic T lymphocytes on HLA‐A2 melanomas. J Exp Med 1993; 178:489–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Coulie PG, Brichard V, Van Pel A, Wolfel T, Schneider J, Traversari C et al A new gene coding for a differentiation antigen recognized by autologous cytolytic T lymphocytes on HLA‐A2 melanomas. J Exp Med 1994; 180:35–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. van der Bruggen P, Traversari C, Chomez P, Lurquin C, De Plaen E, Van den Eynde B et al A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science 1991; 254:1643–7. [DOI] [PubMed] [Google Scholar]
- 27. Kobayashi H, Kumai T, Hayashi S, Matsuda Y, Aoki N, Sato K et al A naturally processed HLA‐DR‐bound peptide from the IL‐9 receptor α of HTLV‐1‐transformed T cells serves as a T helper epitope. Cancer Immunol Immunother 2012; 61:2215–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science 2015; 348:69–74. [DOI] [PubMed] [Google Scholar]
- 29. Gubin MM, Artyomov MN, Mardis ER, Schreiber RD. Tumor neoantigens: building a framework for personalized cancer immunotherapy. J Clin Invest 2015; 125:3413–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Battaglia S, Muhitch JB. Unmasking targets of antitumor immunity via high‐throughput antigen profiling. Curr Opin Biotechnol 2016; 42:92–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Zhang W, Barger CJ, Link PA, Mhawech‐Fauceglia P, Miller A, Akers SN et al DNA hypomethylation‐mediated activation of Cancer/Testis Antigen 45 (CT45) genes is associated with disease progression and reduced survival in epithelial ovarian cancer. Epigenetics 2015; 10:736–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Siebenkas C, Chiappinelli KB, Guzzetta AA, Sharma A, Jeschke J, Vatapalli R et al Inhibiting DNA methylation activates cancer testis antigens and expression of the antigen processing and presentation machinery in colon and ovarian cancer cells. PLoS ONE 2017; 12:e0179501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Roman‐Gomez J, Jimenez‐Velasco A, Agirre X, Castillejo JA, Navarro G, San Jose‐Eneriz E et al Epigenetic regulation of human cancer/testis antigen gene, HAGE, in chronic myeloid leukemia. Haematologica 2007; 92:153–62. [DOI] [PubMed] [Google Scholar]
- 34. Chen YT, Scanlan MJ, Sahin U, Tureci O, Gure AO, Tsang S et al A testicular antigen aberrantly expressed in human cancers detected by autologous antibody screening. Proc Natl Acad Sci USA 1997; 94:1914–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Jager E, Chen YT, Drijfhout JW, Karbach J, Ringhoffer M, Jager D et al Simultaneous humoral and cellular immune response against cancer‐testis antigen NY‐ESO‐1: definition of human histocompatibility leukocyte antigen (HLA)‐A2‐binding peptide epitopes. J Exp Med 1998; 187:265–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Boel P, Wildmann C, Sensi ML, Brasseur R, Renauld JC, Coulie P et al BAGE: a new gene encoding an antigen recognized on human melanomas by cytolytic T lymphocytes. Immunity 1995; 2:167–75. [DOI] [PubMed] [Google Scholar]
- 37. Kerkar SP, Wang ZF, Lasota J, Park T, Patel K, Groh E et al MAGE‐A is more highly expressed than NY‐ESO‐1 in a systematic immunohistochemical analysis of 3668 cases. J Immunother 2016; 39:181–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Wang RF, Rosenberg SA. Human tumor antigens for cancer vaccine development. Immunol Rev 1999; 170:85–100. [DOI] [PubMed] [Google Scholar]
- 39. Wang HY, Wang RF. Enhancing cancer immunotherapy by intracellular delivery of cell‐penetrating peptides and stimulation of pattern‐recognition receptor signaling. Adv Immunol 2012; 114:151–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Novellino L, Castelli C, Parmiani G. A listing of human tumor antigens recognized by T cells: March 2004 update. Cancer Immunol Immunother 2005; 54:187–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Vigneron N, Stroobant V, Van den Eynde BJ, van der Bruggen P. Database of T cell‐defined human tumor antigens: the 2013 update. Cancer Immun 2013; 13:15. [PMC free article] [PubMed] [Google Scholar]
- 42. Morgan RA, Chinnasamy N, Abate‐Daga D, Gros A, Robbins PF, Zheng Z et al Cancer regression and neurological toxicity following anti‐MAGE‐A3 TCR gene therapy. J Immunother 2013; 36:133–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Linette GP, Stadtmauer EA, Maus MV, Rapoport AP, Levine BL, Emery L et al Cardiovascular toxicity and titin cross‐reactivity of affinity‐enhanced T cells in myeloma and melanoma. Blood 2013; 122:863–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Odunsi K, Qian F, Matsuzaki J, Mhawech‐Fauceglia P, Andrews C, Hoffman EW et al Vaccination with an NY‐ESO‐1 peptide of HLA class I/II specificities induces integrated humoral and T cell responses in ovarian cancer. Proc Natl Acad Sci USA 2007; 104:12837–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Odunsi K, Jungbluth AA, Stockert E, Qian F, Gnjatic S, Tammela J et al NY‐ESO‐1 and LAGE‐1 cancer‐testis antigens are potential targets for immunotherapy in epithelial ovarian cancer. Cancer Res 2003; 63:6076–83. [PubMed] [Google Scholar]
- 46. Johnson LA, Morgan RA, Dudley ME, Cassard L, Yang JC, Hughes MS et al Gene therapy with human and mouse T‐cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood 2009; 114:535–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Parkhurst MR, Yang JC, Langan RC, Dudley ME, Nathan DN, Feldman SA et al T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol Ther 2011; 19:620–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines. Nat Med 2004; 10:909–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Lanitis E, Dangaj D, Hagemann IS, Song DG, Best A, Sandaltzopoulos R et al Primary human ovarian epithelial cancer cells broadly express HER2 at immunologically detectable levels. PLoS ONE 2012; 7:e49829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Hogdall EV, Christensen L, Kjaer SK, Blaakaer J, Bock JE, Glud E et al Distribution of HER‐2 overexpression in ovarian carcinoma tissue and its prognostic value in patients with ovarian carcinoma: from the Danish MALOVA Ovarian Cancer Study. Cancer 2003; 98:66–73. [DOI] [PubMed] [Google Scholar]
- 51. Wong YF, Cheung TH, Lam SK, Lu HJ, Zhuang YL, Chan MY et al Prevalence and significance of HER‐2/neu amplification in epithelial ovarian cancer. Gynecol Obstet Invest 1995; 40:209–12. [DOI] [PubMed] [Google Scholar]
- 52. Verri E, Guglielmini P, Puntoni M, Perdelli L, Papadia A, Lorenzi P et al HER2/neu oncoprotein overexpression in epithelial ovarian cancer: evaluation of its prevalence and prognostic significance. Clinical study. Oncology 2005; 68:154–61. [DOI] [PubMed] [Google Scholar]
- 53. Lanitis E, Smith JB, Dangaj D, Flingai S, Poussin M, Xu S et al A human ErbB2‐specific T‐cell receptor confers potent antitumor effector functions in genetically engineered primary cytotoxic lymphocytes. Hum Gene Ther 2014; 25:730–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Long YY, Wang Y, Huang QR, Zheng GS, Jiao SC. Measurement of serum antibodies against NY‐ESO‐1 by ELISA: a guide for the treatment of specific immunotherapy for patients with advanced colorectal cancer. Exp Ther Med 2014; 8:1279–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Fujiwara S, Wada H, Kawada J, Kawabata R, Takahashi T, Fujita J et al NY‐ESO‐1 antibody as a novel tumour marker of gastric cancer. Br J Cancer 2013; 108:1119–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Akcakanat A, Kanda T, Koyama Y, Watanabe M, Kimura E, Yoshida Y et al NY‐ESO‐1 expression and its serum immunoreactivity in esophageal cancer. Cancer Chemother Pharmacol 2004; 54:95–100. [DOI] [PubMed] [Google Scholar]
- 57. Szender JB, Eng KH, Matsuzaki J, Miliotto A, Gnjatic S, Tsuji T et al HLA superfamily assignment is a predictor of immune response to cancer testis antigens and survival in ovarian cancer. Gynecol Oncol 2016; 142:158–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Gnjatic S, Ritter E, Buchler MW, Giese NA, Brors B, Frei C et al Seromic profiling of ovarian and pancreatic cancer. Proc Natl Acad Sci USA 2010; 107:5088–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Oi S, Natsume A, Ito M, Kondo Y, Shimato S, Maeda Y et al Synergistic induction of NY‐ESO‐1 antigen expression by a novel histone deacetylase inhibitor, valproic acid, with 5‐aza‐2’‐deoxycytidine in glioma cells. J Neurooncol 2009; 92:15–22. [DOI] [PubMed] [Google Scholar]
- 60. Klar AS, Gopinadh J, Kleber S, Wadle A, Renner C. Treatment with 5‐Aza‐2’‐deoxycytidine induces expression of NY‐ESO‐1 and facilitates cytotoxic T lymphocyte‐mediated tumor cell killing. PLoS ONE 2015; 10:e0139221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Natsume A, Wakabayashi T, Tsujimura K, Shimato S, Ito M, Kuzushima K et al The DNA demethylating agent 5‐aza‐2’‐deoxycytidine activates NY‐ESO‐1 antigenicity in orthotopic human glioma. Int J Cancer 2008; 122:2542–53. [DOI] [PubMed] [Google Scholar]
- 62. Odunsi K, Matsuzaki J, James SR, Mhawech‐Fauceglia P, Tsuji T, Miller A et al Epigenetic potentiation of NY‐ESO‐1 vaccine therapy in human ovarian cancer. Cancer Immunol Res 2014; 2:37–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Matsuzaki J, Gnjatic S, Mhawech‐Fauceglia P, Beck A, Miller A, Tsuji T et al Tumor‐infiltrating NY‐ESO‐1‐specific CD8+ T cells are negatively regulated by LAG‐3 and PD‐1 in human ovarian cancer. Proc Natl Acad Sci USA 2010; 107:7875–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. van der Bruggen P, Traversari C, Chomez P, Lurquin C, De Plaen E, Van den Eynde BJ et al A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. J Immunol 2007; 178:2617–21. [PubMed] [Google Scholar]
- 65. Markert S, Lassmann S, Gabriel B, Klar M, Werner M, Gitsch G et al α‐Folate receptor expression in epithelial ovarian carcinoma and non‐neoplastic ovarian tissue. Anticancer Res 2008; 28:3567–72. [PubMed] [Google Scholar]
- 66. Chen YL, Chang MC, Huang CY, Chiang YC, Lin HW, Chen CA et al Serous ovarian carcinoma patients with high α‐folate receptor had reducing survival and cytotoxic chemo‐response. Mol Oncol 2012; 6:360–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Canevari S, Mezzanzanica D, Mazzoni A, Negri DR, Ramakrishna V, Bolhuis RL et al Bispecific antibody targeted T cell therapy of ovarian cancer: clinical results and future directions. J Hematother 1995; 4:423–7. [DOI] [PubMed] [Google Scholar]
- 68. Kandalaft LE, Powell DJ Jr, Coukos G. A phase I clinical trial of adoptive transfer of folate receptor‐α redirected autologous T cells for recurrent ovarian cancer. J Transl Med 2012; 10:157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Kershaw MH, Westwood JA, Parker LL, Wang G, Eshhar Z, Mavroukakis SA et al A phase I study on adoptive immunotherapy using gene‐modified T cells for ovarian cancer. Clin Cancer Res 2006; 12:6106–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Song DG, Ye Q, Carpenito C, Poussin M, Wang LP, Ji C et al In vivo persistence, tumor localization, and antitumor activity of CAR‐engineered T cells is enhanced by costimulatory signaling through CD137 (4‐1BB). Cancer Res 2011; 71:4617–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. He X, Wang L, Riedel H, Wang K, Yang Y, Dinu CZ et al Mesothelin promotes epithelial‐to‐mesenchymal transition and tumorigenicity of human lung cancer and mesothelioma cells. Mol Cancer 2017; 16:63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Hassan R, Kreitman RJ, Pastan I, Willingham MC. Localization of mesothelin in epithelial ovarian cancer. Appl Immunohistochem Mol Morphol 2005; 13:243–7. [DOI] [PubMed] [Google Scholar]
- 73. Cheng WF, Huang CY, Chang MC, Hu YH, Chiang YC, Chen YL et al High mesothelin correlates with chemoresistance and poor survival in epithelial ovarian carcinoma. Br J Cancer 2009; 100:1144–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Carpenito C, Milone MC, Hassan R, Simonet JC, Lakhal M, Suhoski MM et al Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains. Proc Natl Acad Sci USA 2009; 106:3360–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Tanyi JL, Haas AR, Beatty GL, Stashwick CJ, O'Hara MH, Morgan MA et al Anti‐mesothelin chimeric antigen receptor T cells in patients with epithelial ovarian cancer. J Clin Oncol 2016; 34:5511. [Google Scholar]
- 76. Lanitis E, Poussin M, Hagemann IS, Coukos G, Sandaltzopoulos R, Scholler N et al Redirected antitumor activity of primary human lymphocytes transduced with a fully human anti‐mesothelin chimeric receptor. Mol Ther 2012; 20:633–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. De Plaen E, Arden K, Traversari C, Gaforio JJ, Szikora JP, De Smet C et al Structure, chromosomal localization, and expression of 12 genes of the MAGE family. Immunogenetics 1994; 40:360–9. [DOI] [PubMed] [Google Scholar]
- 78. Jungbluth AA, Busam KJ, Kolb D, Iversen K, Coplan K, Chen YT et al Expression of MAGE‐antigens in normal tissues and cancer. Int J Cancer 2000; 85:460–5. [PubMed] [Google Scholar]
- 79. Simpson AJ, Caballero OL, Jungbluth A, Chen YT, Old LJ. Cancer/testis antigens, gametogenesis and cancer. Nat Rev Cancer 2005; 5:615–25. [DOI] [PubMed] [Google Scholar]
- 80. Yakirevich E, Sabo E, Lavie O, Mazareb S, Spagnoli GC, Resnick MB. Expression of the MAGE‐A4 and NY‐ESO‐1 cancer‐testis antigens in serous ovarian neoplasms. Clin Cancer Res 2003; 9:6453–60. [PubMed] [Google Scholar]
- 81. Daudi S, Eng KH, Mhawech‐Fauceglia P, Morrison C, Miliotto A, Beck A et al Expression and immune responses to MAGE antigens predict survival in epithelial ovarian cancer. PLoS ONE 2014; 9:e104099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Fritsche HA, Bast RC. CA 125 in ovarian cancer: advances and controversy. Clin Chem 1998; 44:1379–80. [PubMed] [Google Scholar]
- 83. Singh AP, Senapati S, Ponnusamy MP, Jain M, Lele SM, Davis JS et al Clinical potential of mucins in diagnosis, prognosis, and therapy of ovarian cancer. Lancet Oncol 2008; 9:1076–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Chekmasova AA, Rao TD, Nikhamin Y, Park KJ, Levine DA, Spriggs DR et al Successful eradication of established peritoneal ovarian tumors in SCID‐Beige mice following adoptive transfer of T cells genetically targeted to the MUC16 antigen. Clin Cancer Res 2010; 16:3594–606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Koneru M, O'Cearbhaill R, Pendharkar S, Spriggs DR, Brentjens RJ. A phase I clinical trial of adoptive T cell therapy using IL‐12 secreting MUC‐16(ecto) directed chimeric antigen receptors for recurrent ovarian cancer. J Transl Med 2015; 13:102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Urbanska K, Powell DJ Jr. Advances and prospects in adoptive cell transfer therapy for ovarian cancer. Immunotherapy 2015; 7:473–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Algarra I, Cabrera T, Garrido F. The HLA crossroad in tumor immunology. Hum Immunol 2000; 61:65–73. [DOI] [PubMed] [Google Scholar]
- 88. Mandelboim O, Vadai E, Fridkin M, Katz‐Hillel A, Feldman M, Berke G et al Regression of established murine carcinoma metastases following vaccination with tumour‐associated antigen peptides. Nat Med 1995; 1:1179–83. [DOI] [PubMed] [Google Scholar]
- 89. Jurtz V, Paul S, Andreatta M, Marcatili P, Peters B, Nielsen M. NetMHCpan‐4.0: improved peptide‐MHC class I interaction predictions integrating eluted ligand and peptide binding affinity data. J Immunol 2017; 199:3360–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Nielsen M, Andreatta M. NetMHCpan‐3.0; improved prediction of binding to MHC class I molecules integrating information from multiple receptor and peptide length datasets. Genome Med 2016; 8:33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Hoof I, Peters B, Sidney J, Pedersen LE, Sette A, Lund O et al NetMHCpan, a method for MHC class I binding prediction beyond humans. Immunogenetics 2009; 61:1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Andreatta M, Nielsen M. Gapped sequence alignment using artificial neural networks: application to the MHC class I system. Bioinformatics 2016; 32:511–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Nielsen M, Lundegaard C, Worning P, Lauemoller SL, Lamberth K, Buus S et al Reliable prediction of T‐cell epitopes using neural networks with novel sequence representations. Protein Sci 2003; 12:1007–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Nielsen M, Lund O. NN‐align. An artificial neural network‐based alignment algorithm for MHC class II peptide binding prediction. BMC Bioinformatics 2009; 10:296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Nielsen M, Lundegaard C, Lund O. Prediction of MHC class II binding affinity using SMM‐align, a novel stabilization matrix alignment method. BMC Bioinformatics 2007; 8:238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Zhang H, Lund O, Nielsen M. The PickPocket method for predicting binding specificities for receptors based on receptor pocket similarities: application to MHC‐peptide binding. Bioinformatics 2009; 25:1293–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Liu G, Li D, Li Z, Qiu S, Li W, Chao CC et al PSSMHCpan: a novel PSSM‐based software for predicting class I peptide‐HLA binding affinity. Gigascience 2017; 6:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Vita R, Overton JA, Greenbaum JA, Ponomarenko J, Clark JD, Cantrell JR et al The immune epitope database (IEDB) 3.0. Nucleic Acids Res 2015; 43:D405–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. O'Donnell T, Rubinsteyn A, Bonsack M, Riemer A, Hammerbacher J. MHCflurry: open‐source class I MHC binding affinity prediction. bioRxiv 2017; 10.1101/174243 [DOI] [PubMed] [Google Scholar]
- 100. Rammensee H, Bachmann J, Emmerich NP, Bachor OA, Stevanovic S. SYFPEITHI: database for MHC ligands and peptide motifs. Immunogenetics 1999; 50:213–9. [DOI] [PubMed] [Google Scholar]
- 101. Stone JD, Harris DT, Kranz DM. TCR affinity for p/MHC formed by tumor antigens that are self‐proteins: impact on efficacy and toxicity. Curr Opin Immunol 2015; 33:16–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Cohen CJ, Gartner JJ, Horovitz‐Fried M, Shamalov K, Trebska‐McGowan K, Bliskovsky VV et al Isolation of neoantigen‐specific T cells from tumor and peripheral lymphocytes. J Clin Invest 2015; 125:3981–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Martin SD, Coukos G, Holt RA, Nelson BH. Targeting the undruggable: immunotherapy meets personalized oncology in the genomic era. Ann Oncol 2015; 26:2367–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Stronen E, Toebes M, Kelderman S, van Buuren MM, Yang W, van Rooij N et al Targeting of cancer neoantigens with donor‐derived T cell receptor repertoires. Science 2016; 352:1337–41. [DOI] [PubMed] [Google Scholar]
- 105. Pasetto A, Gros A, Robbins PF, Deniger DC, Prickett TD, Matus‐Nicodemos R et al Tumor‐ and neoantigen‐reactive T‐cell receptors can be identified based on their frequency in fresh tumor. Cancer Immunol Res 2016; 4:734–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Gros A, Parkhurst MR, Tran E, Pasetto A, Robbins PF, Ilyas S et al Prospective identification of neoantigen‐specific lymphocytes in the peripheral blood of melanoma patients. Nat Med 2016; 22:433–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. van Rooij N, van Buuren MM, Philips D, Velds A, Toebes M, Heemskerk B et al Tumor exome analysis reveals neoantigen‐specific T‐cell reactivity in an ipilimumab‐responsive melanoma. J Clin Oncol 2013; 31:e439–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Hartmaier RJ, Charo J, Fabrizio D, Goldberg ME, Albacker LA, Pao W et al Genomic analysis of 63,220 tumors reveals insights into tumor uniqueness and targeted cancer immunotherapy strategies. Genome Med 2017; 9:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Hundal J, Carreno BM, Petti AA, Linette GP, Griffith OL, Mardis ER et al pVAC‐Seq: a genome‐guided in silico approach to identifying tumor neoantigens. Genome Med 2016; 8:11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Donnes P, Kohlbacher O. Integrated modeling of the major events in the MHC class I antigen processing pathway. Protein Sci 2005; 14:2132–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Bassani‐Sternberg M, Braunlein E, Klar R, Engleitner T, Sinitcyn P, Audehm S et al Direct identification of clinically relevant neoepitopes presented on native human melanoma tissue by mass spectrometry. Nat Commun 2016; 7:13404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Yadav M, Jhunjhunwala S, Phung QT, Lupardus P, Tanguay J, Bumbaca S et al Predicting immunogenic tumour mutations by combining mass spectrometry and exome sequencing. Nature 2014; 515:572–6. [DOI] [PubMed] [Google Scholar]
- 113. McGranahan N, Furness AJ, Rosenthal R, Ramskov S, Lyngaa R, Saini SK et al Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016; 351:1463–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Tran E, Turcotte S, Gros A, Robbins PF, Lu YC, Dudley ME et al Cancer immunotherapy based on mutation‐specific CD4+ T cells in a patient with epithelial cancer. Science 2014; 344:641–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Klebanoff CA, Rosenberg SA, Restifo NP. Prospects for gene‐engineered T cell immunotherapy for solid cancers. Nat Med 2016; 22:26–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
